Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Stem Cell Res ; 74: 103294, 2024 02.
Article in English | MEDLINE | ID: mdl-38183888

ABSTRACT

BCL2-Associated Athanogene 3 (BAG3) gene was identified mutated in patients with dilated cardiomyopathy (DCM), an important cause of heart failure and premature death. BAG3 is a cytoprotective co-chaperonne protein involved in many cellular process with a central role in the maintenance of protostasis. We generated two human induced pluripotent stem cell lines (hiPSc), one carrying the heterozygous, the other the homozygous p.V468M mutation identified in DCM familial cases. All lines expressed pluripotent markers, had normal karyotype, and differentiated into derivatives of the three germ layers. Sudies of hiPSc derived cardiomyocytes will help to understand the role of BAG3 in DCM.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , CRISPR-Cas Systems/genetics , Apoptosis Regulatory Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Mutation/genetics , Cardiomyopathy, Dilated/genetics
2.
Stem Cell Res ; 60: 102680, 2022 04.
Article in English | MEDLINE | ID: mdl-35093717

ABSTRACT

Mutations leading to haploinsufficiency in SCN5A, the gene encoding the cardiac sodium channel Nav1.5 α-subunit, are involved in life-threatening cardiac disorders. Using CRISPR/Cas9-mediated genome edition, we generated here a human induced-pluripotent stem cell (hiPSC) line carrying a heterozygous mutation in exon 2 of SCN5A, which leads to apparition of a premature stop codon. SCN5A-clone 5 line maintained normal karyotype, morphology and pluripotency and differentiated into three germ layers. Cardiomyocytes derived from these hiPSCs would be a useful model for investigating channelopathies related to SCN5A heterozygous deficiency.


Subject(s)
Induced Pluripotent Stem Cells , CRISPR-Cas Systems/genetics , Heterozygote , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism
3.
Stem Cell Res ; 58: 102616, 2022 01.
Article in English | MEDLINE | ID: mdl-34883448

ABSTRACT

Loss-of-function (LoF) mutations in FLNC are strongly associated with dilated cardiomyopathy (DCM). Using CRISPR/Cas9 mediated edition in an healthy donor derived iPSC (ICAN-403.3) we subcloned 1 iPSC line harboring LoF mutation in FLNC. All lines are fully pluripotent and isogenic except at edited site where it presents a homozygous (ICAN-FLNC42.1) deletion of splice site leading to skipping of exon 42 traduced into a short filamin form with reduced expression in derived cardiomyocytes. This line would serve for FLNC mutation DCM modeling after differentiation into cardiocytes or beating organoids.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , CRISPR-Cas Systems/genetics , Cardiomyopathy, Dilated/genetics , Exons/genetics , Filamins/genetics , Filamins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation
4.
Stem Cell Res ; 52: 102245, 2021 04.
Article in English | MEDLINE | ID: mdl-33610018

ABSTRACT

MYH7 is a major gene responsible for hypertrophic cardiomyopathy (HCM). From patient's skin fibroblasts, we derived an iPSC line (CDGEN1.16) harboring the heterozygous MYH7 R403L mutation, a hot-spot codon in HCM. We subsequently corrected the mutated codon using CRISPR/Cas9 editing and obtained the isogenic control line (CDGEN1.16.40.5) preserving the genomic background of the patient. Both lines were pluripotent and could be efficiently committed to beating cardiomyocytes (CM) suitable for subsequent cell or pseudo-tissue study of HCM pathology.


Subject(s)
Cardiomyopathy, Hypertrophic , Induced Pluripotent Stem Cells , CRISPR-Cas Systems/genetics , Cardiac Myosins/genetics , Cardiomyopathy, Hypertrophic/genetics , Humans , Mutation , Myocytes, Cardiac , Myosin Heavy Chains/genetics
5.
J Am Heart Assoc ; 5(9)2016 09 13.
Article in English | MEDLINE | ID: mdl-27625342

ABSTRACT

BACKGROUND: Mutations in the coding sequence of SCN5A, which encodes the cardiac Na(+) channel α subunit, have been associated with inherited susceptibility to various arrhythmias. Variable expression of SCN5A is a possible mechanism responsible for this pleiotropic effect; however, it is unknown whether variants in the promoter and regulatory regions of SCN5A also modulate the risk of arrhythmias. METHODS AND RESULTS: We resequenced the core promoter region of SCN5A and the regulatory regions of SCN5A transcription in 1298 patients with arrhythmia phenotypes (atrial fibrillation, n=444; sinus node dysfunction, n=49; conduction disease, n=133; Brugada syndrome, n=583; and idiopathic ventricular fibrillation, n=89). We identified 26 novel rare variants in the SCN5A promoter in 29 patients affected by various arrhythmias (atrial fibrillation, n=6; sinus node dysfunction, n=1; conduction disease, n=3; Brugada syndrome, n=14; idiopathic ventricular fibrillation, n=5). The frequency of rare variants was higher in patients with arrhythmias than in controls. In the alignment with chromatin immunoprecipitation sequencing data, the majority of variants were located at regions bound by transcription factors. Using a luciferase reporter assay, 6 variants (Brugada syndrome, n=3; idiopathic ventricular fibrillation, n=2; conduction disease, n=1) were functionally characterized, and each displayed decreased promoter activity compared with the wild-type sequences. We also identified rare variants in the regulatory region that were associated with atrial fibrillation, and the variant decreased promoter activity. CONCLUSIONS: Variants in the core promoter region and the transcription regulatory region of SCN5A were identified in multiple arrhythmia phenotypes, consistent with the idea that altered SCN5A transcription levels modulate susceptibility to arrhythmias.


Subject(s)
Atrial Fibrillation/genetics , Brugada Syndrome/genetics , Cardiac Conduction System Disease/genetics , NAV1.5 Voltage-Gated Sodium Channel/genetics , Sick Sinus Syndrome/genetics , Ventricular Fibrillation/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Arrhythmias, Cardiac/genetics , Child , Female , Genetic Predisposition to Disease , Genetic Variation , Humans , Male , Middle Aged , Mutation , Phenotype , Promoter Regions, Genetic/genetics , Young Adult
6.
ISME J ; 10(1): 51-63, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26114888

ABSTRACT

Like most eukaryotes, brown algae live in association with bacterial communities that frequently have beneficial effects on their development. Ectocarpus is a genus of small filamentous brown algae, which comprises a strain that has recently colonized freshwater, a rare transition in this lineage. We generated an inventory of bacteria in Ectocarpus cultures and examined the effect they have on acclimation to an environmental change, that is, the transition from seawater to freshwater medium. Our results demonstrate that Ectocarpus depends on bacteria for this transition: cultures that have been deprived of their associated microbiome do not survive a transfer to freshwater, but restoring their microflora also restores the capacity to acclimate to this change. Furthermore, the transition between the two culture media strongly affects the bacterial community composition. Examining a range of other closely related algal strains, we observed that the presence of two bacterial operational taxonomic units correlated significantly with an increase in low salinity tolerance of the algal culture. Despite differences in the community composition, no indications were found for functional differences in the bacterial metagenomes predicted to be associated with algae in the salinities tested, suggesting functional redundancy in the associated bacterial community. Our study provides an example of how microbial communities may impact the acclimation and physiological response of algae to different environments, and thus possibly act as facilitators of speciation. It paves the way for functional examinations of the underlying host-microbe interactions, both in controlled laboratory and natural conditions.


Subject(s)
Acclimatization/physiology , Fresh Water/microbiology , Microbial Interactions/physiology , Phaeophyceae/microbiology , Seawater/microbiology , Bacteria/genetics , Metagenome , Microbial Interactions/genetics , Microbiota/genetics , Phaeophyceae/physiology , Salt Tolerance/genetics , Salt Tolerance/physiology
7.
Hum Mol Genet ; 24(10): 2757-63, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25650408

ABSTRACT

The Brugada syndrome (BrS) is a rare heritable cardiac arrhythmia disorder associated with ventricular fibrillation and sudden cardiac death. Mutations in the SCN5A gene have been causally related to BrS in 20-30% of cases. Twenty other genes have been described as involved in BrS, but their overall contribution to disease prevalence is still unclear. This study aims to estimate the burden of rare coding variation in arrhythmia-susceptibility genes among a large group of patients with BrS. We have developed a custom kit to capture and sequence the coding regions of 45 previously reported arrhythmia-susceptibility genes and applied this kit to 167 index cases presenting with a Brugada pattern on the electrocardiogram as well as 167 individuals aged over 65-year old and showing no history of cardiac arrhythmia. By applying burden tests, a significant enrichment in rare coding variation (with a minor allele frequency below 0.1%) was observed only for SCN5A, with rare coding variants carried by 20.4% of cases with BrS versus 2.4% of control individuals (P = 1.4 × 10(-7)). No significant enrichment was observed for any other arrhythmia-susceptibility gene, including SCN10A and CACNA1C. These results indicate that, except for SCN5A, rare coding variation in previously reported arrhythmia-susceptibility genes do not contribute significantly to the occurrence of BrS in a population with European ancestry. Extreme caution should thus be taken when interpreting genetic variation in molecular diagnostic setting, since rare coding variants were observed in a similar extent among cases versus controls, for most previously reported BrS-susceptibility genes.


Subject(s)
Brugada Syndrome/genetics , Genetic Predisposition to Disease , Mutation , NAV1.5 Voltage-Gated Sodium Channel/genetics , Adult , Arrhythmias, Cardiac/genetics , Brugada Syndrome/diagnosis , Female , Gene Frequency , Genes , Genetic Association Studies , Humans , Male , Middle Aged , Sequence Analysis, DNA , White People
8.
Am J Med Genet A ; 158A(7): 1633-40, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22678713

ABSTRACT

Patients with a submicroscopic deletion at 1q43q44 present with intellectual disability (ID), microcephaly, craniofacial anomalies, seizures, limb anomalies, and corpus callosum abnormalities. However, the precise relationship between most of deleted genes and the clinical features in these patients still remains unclear. We studied 11 unrelated patients with 1q44 microdeletion. We showed that the deletions occurred de novo in all patients for whom both parents' DNA was available (10/11). All patients presented with moderate to severe ID, seizures and non-specific craniofacial anomalies. By oligoarray-based comparative genomic hybridization (aCGH) covering the 1q44 region at a high resolution, we obtained a critical deleted region containing two coding genes-HNRNPU and FAM36A-and one non-coding gene-NCRNA00201. All three genes were expressed in different normal human tissues, including in human brain, with highest expression levels in the cerebellum. Mutational screening of the HNRNPU and FAM36A genes in 191 patients with unexplained isolated ID did not reveal any deleterious mutations while the NCRNA00201 non-coding gene was not analyzed. Nine of the 11 patients did not present with microcephaly or corpus callosum abnormalities and carried a small deletion containing HNRNPU, FAM36A, and NCRNA00201 but not AKT3 and ZNF238, two centromeric genes. These results suggest that HNRNPU, FAM36A, and NCRNA00201 are not major genes for microcephaly and corpus callosum abnormalities but are good candidates for ID and seizures.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 1 , Heterogeneous-Nuclear Ribonucleoprotein U/genetics , Intellectual Disability/genetics , RNA, Untranslated/genetics , Seizures/genetics , Child, Preschool , Comparative Genomic Hybridization , Facies , Female , Gene Expression Profiling , Humans , Infant , Karyotyping , Male , Mutation
9.
Eur Heart J ; 32(9): 1065-76, 2011 May.
Article in English | MEDLINE | ID: mdl-21459883

ABSTRACT

AIMS: Dilated cardiomyopathy (DCM) is a major cause of heart failure with a high familial recurrence risk. So far, the genetics of DCM remains largely unresolved. We conducted the first genome-wide association study (GWAS) to identify loci contributing to sporadic DCM. METHODS AND RESULTS: One thousand one hundred and seventy-nine DCM patients and 1108 controls contributed to the discovery phase. Pools of DNA stratified on disease status, population, age, and gender were constituted and used for testing association of DCM with 517 382 single nucleotide polymorphisms (SNPs). Three DCM-associated SNPs were confirmed by individual genotyping (P < 5.0 10(-7)), and two of them, rs10927875 and rs2234962, were replicated in independent samples (1165 DCM patients and 1302 controls), with P-values of 0.002 and 0.009, respectively. rs10927875 maps to a region on chromosome 1p36.13 which encompasses several genes among which HSPB7 has been formerly suggested to be implicated in DCM. The second identified locus involves rs2234962, a non-synonymous SNP (c.T757C, p. C151R) located within the sequence of BAG3 on chromosome 10q26. To assess whether coding mutations of BAG3 might cause monogenic forms of the disease, we sequenced BAG3 exons in 168 independent index cases diagnosed with familial DCM and identified four truncating and two missense mutations. Each mutation was heterozygous, present in all genotyped relatives affected by the disease and absent in a control group of 347 healthy individuals, strongly suggesting that these mutations are causing the disease. CONCLUSION: This GWAS identified two loci involved in sporadic DCM, one of them probably implicates BAG3. Our results show that rare mutations in BAG3 contribute to monogenic forms of the disease, while common variant(s) in the same gene are implicated in sporadic DCM.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cardiomyopathy, Dilated/genetics , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 1/genetics , Genetic Loci/genetics , Heart Failure/genetics , Adult , Apoptosis Regulatory Proteins , Chloride Channels/genetics , Female , Genome-Wide Association Study , HSP27 Heat-Shock Proteins/genetics , Heterozygote , Humans , Male , Middle Aged , Mutation, Missense/genetics , Polymorphism, Single Nucleotide/genetics
10.
Eur Heart J ; 30(17): 2128-36, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19525294

ABSTRACT

AIMS: Dilated cardiomyopathy (DCM) is familial in approximately 30% of cases, and mutations have been identified in several genes. However, in a majority of familial cases, the responsible genes are still to be discovered. The ANKRD1 gene is over-expressed in heart failure in human and animal models. The encoded protein CARP interacts with partners such as myopalladin or titin, previously shown to be involved in DCM. We hypothesized that mutations in ANKRD1 could be responsible for DCM. METHODS AND RESULTS: We sequenced the coding region of ANKRD1 from 231 independent DCM cases. We identified five missense mutations (three sporadic and two familial) absent from 400 controls and affecting highly conserved residues. Expression of the mutant CARP proteins after transfection in rat neonate cardiomyocytes indicated that most of them led to both significantly less repressor activity measured in a reporter gene assay and greater phenylephrin-induced hypertrophy, suggesting altered function of CARP mutant proteins. CONCLUSION: On the basis of genetic and functional analysis of CARP mutations, we have identified ANKRD1 as a new gene associated with DCM, accounting for approximately 2% of cases.


Subject(s)
Cardiomyopathy, Dilated/genetics , Muscle Proteins/genetics , Mutation, Missense/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Adult , Animals , Cardiomyopathy, Hypertrophic/genetics , Cells, Cultured , Female , Gene Expression , Heart Failure/genetics , Heterozygote , Humans , Male , Myocytes, Cardiac , Pedigree , Rats , Transfection
11.
Cardiovasc Res ; 77(1): 118-25, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18006477

ABSTRACT

AIMS: Idiopathic dilated cardiomyopathy (DCM) is a cardiac disorder characterized by left ventricular dilatation and impaired systolic contraction. It is a major cause of heart failure and heart transplantation. DCM is of genetic origin in approximately 30% of cases and genetically heterogeneous with the identification of numerous disease genes. However, many new disease genes remain to be discovered. Focusing on gene products located in the sarcomere of cardiomyocytes as disease-causing candidates, we screened the gene encoding the sarcomeric Z-band protein myopalladin (MYPN, OMIM 608517) for mutation. METHODS AND RESULTS: We sequenced the coding region in 114 (65 familial and 49 sporadic cases) independent DCM patients' DNA and functionally analysed the identified mutations. We identified four independent heterozygous mutations in two families (R1088H and I83fsX105) and two sporadic cases (V1195M, P1112L). For the three missense mutations, the substituted amino acids were conserved among species. All mutations were absent from 400 control subjects. Specific immunolabelling of heart tissue from a proband carrying the R1088H mutation showed a decreased localization of myopalladin at the Z-band area of left ventricular cardiac myofibrils. Analysis of the effects of the mutations after transfection in rat neonate cardiomyocytes indicated sarcomere disorganization and premature cell death associated with the V1195M and P1112L myopalladin expression. Allele-specific expression analysis of mRNA from a patient harbouring the I83fsX105 mutation indicated the absence of the mutated transcript, suggesting a haploinsufficiency mechanism. CONCLUSION: Based on genetic, histological, and functional evidence, we identified a new gene associated with DCM and observed mutations in 3-4% of cases in a population of European descent.


Subject(s)
Cardiomyopathy, Dilated/genetics , Muscle Proteins/genetics , Mutation , Adolescent , Adult , Aged , Amino Acid Sequence , Base Sequence , Cells, Cultured , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Muscle Proteins/chemistry , Polymorphism, Single Nucleotide , RNA, Messenger/analysis
12.
Eur Heart J ; 26(8): 794-803, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15769782

ABSTRACT

AIMS: Familial dilated cardiomyopathy (FDCM) is associated with mutations in more than 10 genes, but genes mutation frequencies and associated clinical features remain largely unknown. Here, we performed a mutation analysis of four genes involved in FDCM in a population of idiopathic DCM. METHODS AND RESULTS: A SSCP and sequencing mutation screening of all the exons coding for beta myosin heavy chain (MYH7 gene), cardiac T troponin (TNNT2 gene), phospholamban (PLN gene), and the cardio-specific exon of metavinculin (VCL gene) were performed in 96 independent patients (54 familial and 42 sporadic). It led to the identification of eight heterozygous mutations, seven new ones in MYH7, and the already described R141W mutation in TNNT2. MYH7 mutations (in five familial and two sporadic cases) substitute residues located either in the head (I201T, T412N, A550V) or tail domains (T1019N, R1193S, E1426K, R1634S) of the protein. DCM was not associated with skeletal myopathy or conduction defects in any patients. Contrasting clinical features were observed between MYH7 and TNNT2 mutations carriers. In MYH7 vs. TNNT2, mean age at diagnosis was late (P<0.03), penetrance was incomplete in adults (56 vs. 100%), and mean age at major cardiac event was higher (P<0.04). CONCLUSION: We have identified seven mutations in MYH7, one in TNNT2, and none in PLN or in the VCL cardio-specific exon. MYH7 appears as the most frequently mutated gene in our FDCM population (approximately 10%), and mutation carriers present with delayed onset, in contrast to TNNT2.


Subject(s)
Cardiomyopathy, Hypertrophic, Familial/genetics , Mutation/genetics , Myosin Heavy Chains/genetics , Adult , Aged , Aged, 80 and over , Child , Female , Genetic Testing/methods , Genotype , Humans , Infant , Male , Middle Aged , Pedigree , Polymorphism, Genetic
13.
Eur J Heart Fail ; 5(6): 821-5, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14675861

ABSTRACT

BACKGROUND: Mutations in LMNA gene encoding two ubiquitously expressed nuclear proteins, lamins A and C, give rise to up to 7 different pathologies affecting specific tissues. Three of these disorders affect cardiac and/or skeletal muscles with atrio-ventricular conduction disturbances, dilated cardiomyopathy and sudden cardiac death as common features. RESULTS: A new LMNA mutation (1621C>T, R541C) was found in two members of a French family with a history of ventricular rhythm disturbances and an uncommon form of systolic left ventricle dysfunction. The two patients: the proband and his daughter, were affected and exhibited an atypical form of dilated cardiomyopathy with an unexplained left ventricle aneurysm revealed by ventricular rhythm disturbances without atrio-ventricular block. CONCLUSION: This finding reinforces the highly variable phenotypic expression of LMNA mutation and emphasizes the fact that LMNA mutations can be associated with different cardiac phenotypes.


Subject(s)
Heart Aneurysm/genetics , Lamin Type A/genetics , Mutation/genetics , Adult , Female , Heart Aneurysm/physiopathology , Humans , Male , Middle Aged , Pedigree , Phenotype , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology
14.
Am J Med Genet A ; 120A(1): 8-12, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12794684

ABSTRACT

Dilated cardiomyopathy (DCM) is defined by ventricular dilatation associated with impaired contractile function. Approximately one-third of idiopathic dilated cardiomyopathy cases are due to inherited gene mutations. Mutations in the beta- and delta-sarcoglycan genes have been described in limb girdle muscular dystrophy and/or isolated DCM. In this study, the aim was to investigate the prevalence of these genes in isolated DCM. We screened these two genes for mutations in 99 unrelated patients with sporadic or familial DCM. The coding exon and intron-exon boundaries of each gene were amplified by polymerase chain reaction. Mutation analyses were performed by single-strand conformation polymorphism for the beta-sarcoglycan gene and by direct sequencing for the delta-sarcoglycan gene. New polymorphisms, as well as already described ones, were found in these two genes, but none appeared to be responsible for dilated cardiomyopathy. We, therefore, conclude that these genes are not responsible for idiopathic isolated dilated cardiomyopathy in our population. Furthermore, based on previously published and present data, we could estimate the prevalence of delta-sarcoglycan gene mutations to be less than 1% in idiopathic dilated cardiomyopathy, demonstrating that this gene is only marginally implicated in the disease.


Subject(s)
Cardiomyopathy, Dilated/genetics , Cytoskeletal Proteins/genetics , DNA Mutational Analysis , Membrane Glycoproteins/genetics , Mutation , Adult , DNA Primers/genetics , Dystroglycans , Exons , Family Health , Female , Humans , Male , Middle Aged , Models, Genetic , Phenotype , Polymorphism, Genetic , Polymorphism, Single-Stranded Conformational , Sarcoglycans
15.
Hum Mutat ; 21(5): 473-81, 2003 May.
Article in English | MEDLINE | ID: mdl-12673789

ABSTRACT

Heritable dilated cardiomyopathy is a genetically highly heterogeneous disease. To date 17 different chromosomal loci have been described for autosomal dominant forms of dilated cardiomyopathy with or without additional clinical manifestations. Among the 10 mutated genes associated with dilated cardiomyopathy, the lamin A/C (LMNA) gene has been reported in forms associated with conduction-system disease with or without skeletal muscle myopathy. For the first time, we report here a French family affected with a new phenotype composed of an autosomal dominant severe dilated cardiomyopathy with conduction defects or atrial/ventricular arrhythmias, and a specific quadriceps muscle myopathy. In all previously reported cases with both cardiac and neuromuscular involvement, neuromuscular disorders preceded cardiac abnormalities. The screening of the coding sequence of the LMNA gene on all family members was performed and we identified a missense mutation (R377H) in the lamin A/C gene that cosegregated with the disease in the family. Cell transfection experiments showed that the R377H mutation leads to mislocalization of both lamin and emerin. These results were obtained in both muscular (C2C12) and non-muscular cells (COS-7). This new phenotype points out the wide spectrum of neuromuscular and cardiac manifestations associated with lamin A/C mutations, with the functional consequence of this mutation seemingly associated with a disorganization of the lamina.


Subject(s)
Cardiomyopathy, Dilated/genetics , Lamin Type A/genetics , Myocardium/pathology , Adult , Animals , COS Cells , Cardiomyopathy, Dilated/pathology , Cell Line , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Desmin/analysis , Dystrophin/analysis , Family Health , Female , Humans , Immunohistochemistry , Lamin Type A/analysis , Male , Membrane Proteins/analysis , Middle Aged , Muscle, Skeletal/chemistry , Muscle, Skeletal/pathology , Mutation , Mutation, Missense , Myocardium/metabolism , Nuclear Proteins , Pedigree , Plasmids/drug effects , Thymopoietins/analysis , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...